Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Paul J. Cannon is active.

Publication


Featured researches published by Paul J. Cannon.


Journal of Clinical Investigation | 1995

The lethal effects of cytokine-induced nitric oxide on cardiac myocytes are blocked by nitric oxide synthase antagonism or transforming growth factor beta.

David J. Pinsky; Bolin Cai; Xiaochun Yang; C. Rodriguez; Robert R. Sciacca; Paul J. Cannon

Inducible nitric oxide (NO) produced by macrophages is cytotoxic to invading organisms and has an important role in host defense. Recent studies have demonstrated inducible NO production within the heart, and that cytokine-induced NO mediates alterations in cardiac contractility, but the cytotoxic potential of nitric oxide with respect to the heart has not been defined. To evaluate the role of inducible nitric oxide synthase (iNOS) on cardiac myocyte cytotoxicity, we exposed adult rat cardiac myocytes to either cytokines alone or to activated J774 macrophages in coculture. Increased expression of both iNOS message and protein was seen in J774 macrophages treated with IFN gamma and LPS and cardiac myocytes treated with TNF-alpha, IL-1 beta, and IFN gamma. Increased NO synthesis was confirmed in both the coculture and isolated myocyte preparations by increased nitrite production. Increased NO synthesis was associated with a parallel increase in myocyte death as measured by CPK release into the culture medium as well as by loss of membrane integrity, visualized by trypan blue staining. Addition of the competitive NO synthase inhibitor L-NMMA to the culture medium prevented both the increased nitrite production and the cytotoxicity observed after cytokine treatment in both the isolated myocyte and the coculture experiments. Because transforming growth-factor beta modulates iNOS expression in other cell types, we evaluated its effects on cardiac myocyte iNOS expression and NO-mediated myocyte cytotoxicity. TGF-beta reduced expression of cardiac myocyte iNOS message and protein, reduced nitrite production, and reduced NO-mediated cytotoxicity in parallel. Taken together, these experiments show the cytotoxic potential of endogenous NO production within the heart, and suggest a role for TGF-beta or NO synthase antagonists to mute these lethal effects. These findings may help explain the cardiac response to sepsis or allograft rejection, as well as the progression of dilated cardiomyopathies of diverse etiologies.


Arteriosclerosis, Thrombosis, and Vascular Biology | 1997

Nitric oxide synthases and cardiac muscle. Autocrine and paracrine influences.

Jean-Luc Balligand; Paul J. Cannon

The different cell types comprising cardiac muscle express one or more of the three isoforms (neuronal NOS, or nNOS; inducible NOS, or iNOS; and endothelial NOS, or eNOS) of nitric oxide synthase (NOS). nNOS is expressed in orthosympathetic nerve terminals and regulates the release of catecholamines in the heart. eNOS constitutively expressed in endothelial cells inhibits contractile tone and the proliferation of underlying vascular smooth muscle cells, inhibits platelet aggregation and monocyte adhesion, promotes diastolic relaxation, and decreases O2 consumption in cardiac muscle through paracrinally produced NO. eNOS is also constitutively expressed in cardiac myocytes from rodent and human species, where it autocrinally opposes the inotropic action of catecholamines after muscarinic cholinergic and beta-adrenergic receptor stimulation. iNOS gene transcription and protein expression are induced in all cell types after exposure to a variety of inflammatory cytokines. Aside from participating in the immune defense against intracellular microorganisms and viruses, the large amounts of NO produced autocrinally or paracrinally mediate the vasoplegia and myocardial depression characteristic of systemic immune stimulation and promote cell death through apoptosis. In cardiac myocytes, NO may regulate L-type calcium current and contraction through activation of cGMP-dependent protein kinase and cGMP-modulated phosphodiesterases. Other mechanisms independent of cGMP elevations may operate through interaction of NO with heme proteins, non-heme iron, or free thiol residues on target signaling proteins, enzymes, or ion channels. Given the multiplicity of NOS isoforms expressed in cardiac muscle and of the potential molecular targets for the NO produced, tight molecular regulation of NOS expression and activity at the transcriptional and posttranscriptional level appear to be needed to coordinate the many roles of NO in heart function in health and disease.


Journal of Clinical Investigation | 1994

Induction of myocardial nitric oxide synthase by cardiac allograft rejection.

Xiaochun Yang; Nepal C. Chowdhury; Bolin Cai; Jerold Brett; Charles C. Marboe; Robert R. Sciacca; Robert E. Michler; Paul J. Cannon

Cardiac transplantation, effective therapy for end-stage heart failure, is frequently complicated by allograft rejection, the mechanisms of which remain incompletely understood. Nitric oxide (NO), a vasodilator which is cytotoxic and negatively inotropic, can be produced in large amounts by an inducible NO synthase (iNOS) in response to cytokines. To investigate whether iNOS is induced during cardiac allograft rejection, hearts from Lewis or Wistar-Furth rats were transplanted into Lewis recipients. At day 5, allogeneic grafts manifested reduced contractility and histologic evidence of rejection (inflammatory infiltrate, edema, necrosis of myocytes). The mRNA for iNOS and iNOS protein were detected in ventricular homogenates and in isolated cardiac myocytes from rejecting allogeneic grafts but not in tissue and myocytes from syngeneic control grafts. Immunocytochemistry showed increased iNOS staining in infiltrating macrophages and in microvascular endothelial cells and cardiac muscle fibers and also in isolated purified cardiac myocytes from the rejecting allografts. Using a myocardial cytosolic iNOS preparation, nitrite formation from L-arginine and [3H] citrulline formation from [3H]L-arginine were increased significantly in the rejecting allogeneic grafts (P < 0.01). Myocardial cyclic GMP was also increased significantly (P < 0.05). The data indicate myocardial iNOS mRNA, protein and enzyme activity are induced in infiltrating macrophages and cardiac myocytes of the rejecting allogeneic grafts. Synthesis of NO by iNOS may contribute to myocyte necrosis and ventricular failure during cardiac allograft rejection.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2000

Acute Elevations of Plasma Asymmetric Dimethylarginine and Impaired Endothelial Function in Response to a High-Fat Meal in Patients With Type 2 Diabetes

Ali Fard; Catherine Tuck; Joshua Donis; Robert R. Sciacca; Marco R. Di Tullio; Henry D. Wu; Todd A. Bryant; Niem Tzu Chen; Margarita Torres-Tamayo; Ravichandran Ramasamy; Lars Berglund; Henry N. Ginsberg; Shunichi Homma; Paul J. Cannon

Asymmetric dimethylarginine (ADMA), a compound detectable in human plasma, is an endogenous inhibitor of NO synthase. Endothelial dysfunction is an early event in atherogenesis, and large-vessel atherosclerosis is a major cause of morbidity and mortality in patients with type 2 diabetes mellitus. Fifty patients with type 2 diabetes mellitus were studied at baseline and 5 hours after ingestion of a high-fat meal. Plasma ADMA measured by using high-performance liquid chromatography increased from 1.04±0.99 to 2.51±2.27 &mgr;mol/L (P <0.0005). Brachial arterial vasodilation after reactive hyperemia, a NO-dependent function, measured by high-resolution ultrasound, decreased from 6.9±3.9% at baseline to 1.3±4.5% (P <0.0001). These changes occurred in association with increased plasma levels of triglycerides and very low density lipoprotein triglycerides, with reduced low density lipoprotein cholesterol and high density lipoprotein cholesterol, and with no changes in total cholesterol. The increase in plasma ADMA in response to a high-fat meal was significantly and inversely related to the decrease in percent vasodilation. In 10 of the subjects studied with a similar protocol on another day, no significant changes in the brachial artery flow responses or in plasma ADMA were observed 5 hours after ingestion of a nonfat isocaloric meal. The data suggest that ADMA may contribute to abnormal blood flow responses and to atherogenesis in type 2 diabetics.


Circulation | 1996

Apoptosis of Cardiac Myocytes During Cardiac Allograft Rejection: Relation to Induction of Nitric Oxide Synthase

Matthias Szabolcs; Robert E. Michler; Xiaochun Yang; Walif Aji; Dilip K. Roy; Eleni Athan; Robert R. Sciacca; Oktavjian P. Minanov; Paul J. Cannon

BACKGROUND Apoptosis is a distinct form of programmed cell death characterized by activation of endonucleases that cleave nuclear DNA, condensation and fragmentation of nuclear chromatin, blebbing of intact membranes, and cell shrinkage and fragmentation. The mechanisms responsible are unclear, but nitric oxide (NO) generated by inducible NO synthase (iNOS) has been demonstrated to induce apoptosis in macrophages in vitro. This study investigated whether apoptosis occurs during cardiac allograft rejection and examined the relationship of apoptosis to iNOS expression. METHODS AND RESULTS Heterotopic abdominal transplantation from Lewis to Wistar-Furth rats was used as a model of cardiac allograft rejection; Lewis-to-Lewis grafts served as controls. Apoptosis was identified by DNA ladders after electrophoresis on agarose gels and by in situ labeling of DNA fragments; cell types were determined by immunohistochemistry. The number of apoptotic cardiac myocytes increased sharply from day 3 (0.31/mm2 ventricular tissue) to day 5 (1.27/mm2) after transplantation. At day 5, allografts showed a significant increase (P < .01) in apoptotic cardiac myocytes, macrophages, and endothelial cells compared with syngeneic grafts. The expression of iNOS mRNA, protein, and enzyme activity paralleled in time and extent the apoptosis of cardiac myocytes. iNOS immunostaining of infiltrating macrophages and cardiac muscle fibers increased significantly in the allografts at days 3 to 5 and was accompanied by immunostaining of both cell types for nitrotyrosine, which is indicative of peroxynitrite formation. CONCLUSIONS Apoptosis of myocardial cells occurs during cardiac allograft rejection. Apoptosis during rejection parallels the expression of iNOS, which suggests that apoptosis may be triggered by NO and peroxynitrite.


Journal of Clinical Investigation | 1963

ANGIOTENSIN II, NOREPINEPHRINE, AND RENAL TRANSPORT OF ELECTROLYTES AND WATER IN NORMAL MAN AND IN CIRRHOSIS WITH ASCITES*

John H. Laragh; Paul J. Cannon; Carl J. Bentzel; Alfred M. Sicinski; Jay I. Meltzer

In normal subjects, the rate of aldosterone secretion fluctuates widely according to the state of salt and water balance, but the pathways mediating this relationship are still to be identified. Hypersecretion of aldosterone occurs in malignant nephrosclerosis (2). Angiotensin, the pressor peptide released from plasma by renin, increases both the adrenal secretion (3) and the urinary excretion (4) of aldosterone in man. These observations support the suggestion (5) that a renaladrenal interaction might be involved in the normal regulation of sodium and potassium balance. By this mechanism, the kidney, perhaps sensitive to diminished arterial filling or ischemia, secretes renin, generating angiotensin, which then stimulates aldosterone secretion. Aldosterone, in turn, restores renal perfusion by promoting sodium and water retention. In malignant hypertension this mechanism may become deranged, leading to the coexistence of increased amounts of both angiotensin and aldosterone in the blood, a possibility that derives additional support from the earlier report (6) of increased blood angiotensin in these patients. On the other hand, since neither renin nor angiotensin has been conclusively demonstrated in normal human plasma, elaboration of these substances in malignant hypertension might be simply a consequence of severe renal damage. Our more recent work has investigated the possibility that this renal-adrenal interaction participates in normal homeostasis, as well as in the


Circulation | 1997

l-Arginine Prevents Xanthoma Development and Inhibits Atherosclerosis in LDL Receptor Knockout Mice

Walif Aji; Stefano Ravalli; Matthias Szabolcs; Xian-cheng Jiang; Robert R. Sciacca; Robert E. Michler; Paul J. Cannon

BACKGROUND The potential antiatherosclerotic actions of NO were investigated in four groups of mice (n = 10 per group) lacking functional LDL receptor genes, an animal model of familial hypercholesterolemia. Group 1 was fed a regular chow diet. Groups 2 through 4 were fed a 1.25% high-cholesterol diet. In addition, group 3 received supplemental L-arginine and group 4 received L-arginine and N omega-nitro-L-arginine (L-NA), an inhibitor of NO synthase (NOS). METHODS AND RESULTS Animals were killed at 6 months; aortas were stained with oil red O for planimetry and with antibodies against constitutive and inducible NOSs. Plasma cholesterol was markedly increased in the animals receiving the high-cholesterol diet. Xanthomas appeared in all mice fed the high-cholesterol diet alone but not in those receiving L-arginine. Aortic atherosclerosis was present in all mice on the high-cholesterol diet. The mean atherosclerotic lesion area was reduced significantly (P < .01) in the cholesterol-fed mice given L-arginine compared with those receiving the high-cholesterol diet alone. The mean atherosclerotic lesion area was significantly larger (P < .01) in cholesterol-fed mice receiving L-arginine + L-NA than in those on the high-cholesterol diet alone. Within the atherosclerotic plaques, endothelial cells immunoreacted for endothelial cell NOS; macrophages, foam cells, and smooth muscle cells immunostained strongly for inducible NOS and nitrotyrosine residues. CONCLUSIONS The data indicate that L-arginine prevents xanthoma formation and reduces atherosclerosis in LDL receptor knockout mice fed a high-cholesterol diet. The abrogation of the beneficial effects of L-arginine by L-NA suggests that the antiatherosclerotic actions of L-arginine are mediated by NOS. The data suggest that L-arginine may be beneficial in familial hypercholesterolemia.


Transplantation | 1998

Apoptosis and increased expression of inducible nitric oxide synthase in human allograft rejection

Matthias Szabolcs; Stefano Ravalli; Oktavijan P. Minanov; Robert R. Sciacca; Robert E. Michler; Paul J. Cannon

BACKGROUND The mechanisms of myocyte death during cardiac allograft rejection are incompletely understood. In a previous study using a rat heterotopic cardiac allograft model, we showed that cardiac myocyte apoptosis, inducible nitric oxide synthase (iNOS) mRNA, protein and enzyme activity, and nitrotyrosine increased simultaneously during cardiac allograft rejection. This study was designed to investigate whether apoptosis and expression of iNOS occur in human cardiac allograft rejection. METHODS Right ventricular endomyocardial biopsies from 30 cases of allograft rejection (International Society of Heart and Lung Transplantation grade 3A/B) were compared with 12 biopsies with no rejection (International Society of Heart and Lung Transplantation grade 0). Samples were co-labeled for apoptosis and muscle actin. Serial sections were stained for iNOS, nitrotyrosine, and the leukocyte markers CD3, CD4, CD8, and CD68 to identify T-cell subpopulations and macrophages. RESULTS Biopsies with cardiac allograft rejection showed a 30-fold increase of apoptotic cells when compared with controls. Most apoptotic cardiac myocytes were found in proximity to macrophage (CD68+)-rich inflammatory infiltrates. iNOS immunoreactivity was strongest in macrophages and adjacent myocytes, which also showed high levels of nitrotyrosine, representing damage by peroxynitrite. CONCLUSIONS Apoptosis is a major form of myocyte death during human cardiac allograft rejection. Cardiac myocyte apoptosis is closely associated with expression of iNOS in macrophages and myocytes and with nitration of myocyte proteins by peroxynitrite.


Journal of the American College of Cardiology | 1989

Antimyosin imaging in acute transmural myocardial infarctions: Results of a multicenter clinical trial

Lynne L. Johnson; David W. Seldin; Lewis C. Becker; Norman LaFrance; Henry A. Liberman; Christopher James; Jeffrey A. Mattis; Richard T. Dean; John W. Brown; Andrew Reiter; Valerie Arneson; Paul J. Cannon; Harvey J. Berger

Murine monoclonal antimyosin antibody has been shown experimentally to bind selectively to irreversibly damaged myocytes. To evaluate the safety and efficacy of monoclonal antimyosin for identifying acute transmural infarction, 50 patients with acute Q wave myocardial infarction were entered into a phase I/II multicenter trial involving three clinical sites. Indium-111 antimyosin was prepared from an instant kit formulation containing 0.5 mg of diethylene triamine pentaacetic acid (DTPA)-coupled Fab fragment (R11D10) and 1.2 to 2.4 mCi of indium-111. Average labeling efficiency was 92%. Antimyosin was injected 27 +/- 16 h after the onset of chest pain. Planar or tomographic imaging was performed 27 +/- 9 h after injection in all patients, and repeat imaging was done 24 h later in 39 patients. Of the 50 patients entered, 46 showed myocardial uptake of antimyosin (sensitivity 92%). Thirty-one of 39 planar scans performed at 24 h were diagnostic; 8 showed persistent blood pool activity that cleared by 48 h. Focal myocardial uptake of antimyosin corresponded to electrocardiographic infarct localization. No patient had an adverse reaction to antimyosin. In addition, 125 serum samples, including 21 collected greater than 42 days after injection, were tested for human antimouse antibodies, and all samples were assessed as having undetectable titers. Intensity of antimyosin uptake was correlated with infarct location and the presence or absence of collateral vessels. There was a significant correlation between faint uptake and inferoposterior infarct location. In 21 patients who had coronary angiography close to the time of antimyosin injection, there was a significant correlation between faint tracer uptake and closed infarct-related vessel with absent collateral flow.(ABSTRACT TRUNCATED AT 250 WORDS)


The New England Journal of Medicine | 1977

The Kidney in Heart Failure

Paul J. Cannon

The kidneys participate to a major extent in the pathogenesis of congestive heart failure. When the heart fails as a pump, a complex alteration of homeostatic mechanisms results. Among the adjustme...

Collaboration


Dive into the Paul J. Cannon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert E. Michler

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge